[go: up one dir, main page]
More Web Proxy on the site http://driver.im/ Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 15.
Published in final edited form as: Cancer Res. 2018 Nov 6;79(2):372–386. doi: 10.1158/0008-5472.CAN-18-1334

Antifibrotic therapy disrupts stromal barriers and modulates the immune landscape in pancreatic ductal adenocarcinoma

Kianna Y Elahi-Gedwillo 1,2, Marjorie Carlson 1, Jon Zettervall 1, Paolo P Provenzano 1,2,3,4,5
PMCID: PMC6335156  NIHMSID: NIHMS1512006  PMID: 30401713

Abstract

Pancreatic ductal adenocarcinoma (PDA) remains one of the deadliest forms of cancer, in part, because it is largely refractory to current therapies. The failure of most standard therapies in PDA, as well as promising immune therapies, may be largely ascribed to highly unique and protective stromal microenvironments that present significant biophysical barriers to effective drug delivery, that are immunosuppressive, and that can limit the distribution and function of anti-tumor immune cells. Here, we utilized stromal re-engineering to disrupt these barriers and move the stroma toward normalization using a potent antifibrotic agent, halofuginone. In an autochthonous genetically engineered mouse model of PDA, halofuginone disrupted physical barriers to effective drug distribution by decreasing fibroblast activation and reducing key extracellular matrix elements that drive stromal resistance. Concomitantly, halofuginone treatment altered the immune landscape in PDA, with greater immune infiltrate into regions of low hylauronan, which resulted in increased number and distribution of both classically activated inflammatory macrophages and cytotoxic T cells. In concert with a direct effect on carcinoma cells, this led to widespread intratumoral necrosis and reduced tumor volume. These data point to the multifunctional and critical role of the stroma in tumor protection and survival and demonstrate how compromising tumor integrity to move toward a more normal physiologic state through stroma-targeting therapy will likely be an instrumental component in treating PDA.

Keywords: Pancreatic cancer, Tumor and immune microenvironment, Drug transport and resistance, Stroma targeting therapy

INTRODUCTION

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal and therapeutically nonresponsive cancers, with nearly identical annual incidence and mortality rate (1). Despite an improved understanding of PDA, 5-year survival has scarcely improved from 5% to 9% (1). Among other factors, including early metastasis and robustly immunosuppressive tumor microenvironments, the difficulty in treating PDA may be largely attributed to protective stromal barriers that effectively limit drug delivery, and distribution of anti-tumor immune cell populations (26). Indeed, PDA exhibits a striking lack of vasculature (3,4), in contrast to virtually all other solid tumors, and extremely elevated interstitial fluid pressure (IFP)(4,7). This pressure gradient contributes to vascular collapse and effectively blocks transvascular convective (i.e. pressure-driven) transport of small molecules from perfused vessels (4,7,8), while dense collagen and glyocosaminoglycan networks also hinder distribution of therapeutics by diffusive (i.e. concentration-driven) transport (4,7,9,10). Collectively, these barriers severely restrict intratumoral drug distribution, resulting in large drug-free regions in PDA.

An overwhelming majority of the dense ECM in PDA is deposited by activated pancreatic stellate cells (PSCs, i.e. myofibroblasts or cancer-associated fibroblasts, CAFs), which can be activated via inflammation, paracrine signaling, and oxidative stress (11,12) and also promote carcinoma cell proliferation, migration, and survival (11,13,14) and inception of metastatic lesions (14). Furthermore, activated PSCs can also promote immunosuppression through cytokine secretion (15,16), and recent evidence suggests that these α-SMA+ CAFs can display significant heterogeneity with some CAFs possessing stronger inflammatory functions (15). Collectively, these results make PSCs and the ECM attractive therapeutic targets in PDA. However, notably, there are conflicting reports regarding the relative benefit or harm of stroma-targeting therapies. Pharmacologic and/or genetic depletion of the stroma by targeting Hedgehog (Hh) signaling in preinvasive disease does not prevent PDA and may actually result in more aggressive disease (17), while separate studies report a benefit from targeting Hh in preclinical models (3,18,19) and human patients (20). Distinct from approaches targeting Hh signaling, other studies have indicated a strong therapeutic benefit from stroma targeting. Enzymatic targeting of hyaluronan in authochonous PDA (4) and angiotensin inhibition in grafted tumors (9,10) lead to disruption of physical barriers, reduced intratumoral pressure, and restored vascular functionality to facilitate drug delivery and, in some cases, long-term stromal remodeling (4). These studies suggest that our understanding of stromal signaling and physical barriers, and how and when to alter the stroma, remains incomplete, and that novel combinatorial molecular and immune approaches are likely needed in concert with an increased understanding of how stromal re-engineering alters pro- and anti-tumor immunity.

We investigate the therapeutic potential of the potent antifibrotic agent halofuginone (HF), an analog of febrifugine, to move the tumor toward stromal normalization by targeting activated PSCs and inhibiting ECM production, thereby disrupting barriers to effective molecular and immune therapies. Febrifugine has been used as an antimalarial and anti-parasitic agent, but more recently discovered to have strong antifibrotic capabilities. While the exact mechanism of action remains somewhat unclear, HF blocks TGF-β signaling through inhibition of Smad2 and Smad3 phosphorylation and can reduce expression TGF-β type II receptor (21), which may result from inhibition of prolyl-tRNA synthetase activity (22). As a result, HF can decrease pancreatitis-induced fibrosis, slow growth of grafted pancreatic tumors (2325), and has elicited resolution of pathologic liver fibrosis (26). Furthermore, HF has been administered safely in human patients with advanced solid tumors (27). Yet, while the antifibrotic effects of HF have been studied in various contexts, they have not been previously tested in an autochthonous model of PDA. Here, we show that HF is able to disrupt physical barriers in established stroma to facilitate increased therapeutic delivery. Notably, HF treatment also promotes infiltration and distribution of anti-tumor immune populations, including classically activated inflammatory macrophages and CD8+ cytotoxic T cells. In concert with a direct effect on carcinoma cells, HF treatment culminates in widespread intratumoral necrosis and reduced tumor volume growth in a genetically engineered mouse model of PDA.

MATERIALS AND METHODS

Cell lines.

All murine primary cell lines were generated from freshly excised KPC tumors (the KPC genetically engineered mouse model is described in detail in following section) in our laboratory and were confirmed to be mycoplasma-free. Primary carcinoma cells were confirmed to have the correct genotype and to have undergone Cre-mediated recombination resulting in activation of the Kras oncogene and point mutant p53. To isolate primary carcinoma cells and activated PSCs (i.e. CAFs), tumors were washed with a 10% penicillin streptomycin solution, minced into small pieces, submerged in 2 mg/ml collagenase and 100U/mL hyaluronidase at 37°C for 30 minutes with agitation, washed and resuspended five times in DMEM, then plated onto FBS-coated tissue culture plates. Pancreatic stellate cells were isolated from cell mixtures by differential trypsinization of mixed populations of cells to highly enrich PSCs, followed by resuspension and incubation with in a biotinylated mouse E-cadherin antibody solution (PlusCellect Mouse E-cadherin+ Cancer/Stem Cells Kit) in combination with anti-β4 integrin and anti-CD11b, followed by resuspension and incubation with Dynabeads Biotin Binder (ThermoFisher Scientific) to remove carcinoma cells and macrophages. Following the initial purification differential trypsinization and selection with only E-cadherin beads was used to maintain high PSC purity as needed. Cells were used between 3–15 passages. Human primary PSCs were generated using an outgrowth method (28). De-identified resectable human PDA was finely minced. Tissues were initially cultured in DMEM (Invitrogen) with 20% FCS (“CP-medium”), then second, in KSF medium (Invitrogen), and third, Dresden modified DME medium (“Dresden-medium”), consisting of CP medium and KSF medium at a ratio of 2:1. All media was supplemented with penicillin (100 U/mL) and gentamicin (2.5 mg/mL)(Invitrogen). Differential trypsinization was implemented to isolate PSCs from human tumors, no carcinoma cells were detected in the culture that were confirmed to be mycoplasma-free. Cells were used within 10 passages. All human samples were de-identified and obtained through the UMN BioNet program in accordance with the US Department of Health and Human Services (HHS) Common Rule and University of Minnesota IRB approval.

Genetically engineered mouse model.

All animal studies were approved by the Institutional Animal Care and Use Committee of the University of Minnesota. In vivo studies utilized the KrasG12D/+;p53R172H/+;Pdx-1-Cre (KPC) genetically engineered mouse model, which has proven to be highly reflective of the human cognate condition genetically, clinically, and histopathologically (29). KPC mice conditionally express endogenous, physiologic levels of the respective oncogene and tumor suppressor gene mutations in pancreatic progenitor cells, resulting in the stochastic development and spontaneous progression of preinvasive ductal lesions to invasive and metastatic carcinomas. A subset of studies were done using fluorescent reporter KPC mice (i.e. KrasG12D/+;p53R172H/+;Pdx-1-Cre;ROSATdtomato/+ (KPCT) or KrasG12D/+;p53R172H/+;Pdx-1-Cre;ZsGreen+/+ (KPCG) as we have previously described (30)).

KPC study endpoint protocol.

At the endpoint of the drug studies, doxorubicin was injected i.v. 15–20 minutes prior to euthanasia to visualize small molecule distribution in tumors. This was followed by a terminal surgery, in which animals were anaesthetized using isofluorane and an intracardiac (i.c.) injection of high molecular weight (2000 kDa) FITC- or TRITC-conjugated lysine fixable dextran was administered 5 minutes prior to euthanasia to label perfused blood vessels, followed by a terminal i.c. injection of 10% formalin. The organs were harvested and fixed in 10% neutral buffered formalin, followed by dehydration in 70% ethanol before paraffin-embedding and sectioning (4 μm) for subsequent histochemical or immunohistochemical staining. Separate sections of each sample were frozen in O.C.T. compound.

Ultrasound for enrollment and monitoring of disease progression.

Weekly ultrasound was performed on KPC mice using the Vevo® 2100 Imaging System for both enrollment and disease monitoring. When tumors reached a diameter of 4–9mm, mice were randomly enrolled in study groups. The Vevo® 2100 software was used to reconstruct 3-D tumor volumes for quantification of tumor volume growth over time.

Drug treatment plans and dosages.

Unless specifically noted, the mice described in this study were enrolled in 10-day intervention studies using halofuginone, an analog of the plant alkaloid, febrifugine. A small subset of mice were enrolled in 24-hour intervention studies to evaluate early immune dynamics following one dose of 0.75 mg/kg halofuginone hydrobromide (Sigma Aldrich) administered intraperitoneal (i.p.), followed by euthanasia 24 hours later (n=3). Mice enrolled in the control group received no treatment, but were continually monitored, then euthanized after 10 days (n=6). Mice in the 10-day HF group were treated 3x/week, receiving a total of 5 doses (n=5).

Measuring interstitial fluid pressure.

Tumor pressures were measured as previously described (3). Briefly, interstitial fluid pressure (IFP) measurements were performed using a Millar Mikro-Tip system (pressure catheter transducer SPR-1000, 0.33 mm diameter) connected to a PCU-2000 Pressure Control Unit and the ADInstruments PowerLab data acquisition system (Millar Instruments, Inc.) The system was calibrated to 0, 25, and 100 mm Hg prior to each measurement following the manufacturer’s recommendations.

Histological analysis.

All stained tissue samples were digitally scanned at high resolution and viewed using Aperio ImageScope. Using this software we identified PDA-positive area within H&E stained sections. Likewise, this approach was used to identify and quantify necrotic area as a percentage of total PDA area. To quantify IHC stained tissue sections, ten regions of interest were randomly selected within the PDA-positive area, defined by referencing previously identified PDA-positive areas in adjacent H&E stained sections. Macros were written in FIJI to analyze each histological stain, using batch image processing to isolate and quantify the relevant positive signal. To identify perfused blood vessels, 2000 kDa FITC- (ThermoFisher Scientific) or TRITC-conjugated (Life Technologies) lysine-fixable dextrans were injected intracardiac (i.c.) 5 minutes prior to death, followed by a terminal i.c. injection of 10% formalin. Perfused blood vessels were imaged using an inverted Olympus IX81ZDC spinning disk confocal microscope. Corresponding regions were identified in adjacent tissue slices stained for CD31 and used to calculate total number of vessels. Percent perfusion was calculated as the number of fluorescently labeled, perfused blood vessels as a percent of the total number of CD31+ vessels in that region. To estimate immune infiltrate in H&E sections, a quantitative image-processing algorithm was created in FIJI. A built-in color deconvolution plugin was used to separate the H&E color channels and isolate the nuclei. Immune cells were distinguished by thresholding for specific nuclear characteristics (immune cells were presumed to have very small, round, and dark nuclei). Nuclear size parameters were determined by iterating the pixel range until the maximum particle size was small enough to exclude the visibly larger nuclei of carcinoma cells, and the minimum was small enough to exclude random debris. Circularity was set to 0.75–1.00 to exclude the elongated nuclei of CAFs. An example of the algorithm’s input and output is included in Figure S4.

Immunohistochemistry staining.

Formalin-fixed paraffin-embedded (FFPE) tissue sections were stained for αSMA (1:25; Dako, M0851), CD31 (1:50; Dianova, DIA310), iNos (1:150 Abcam; ab15323) and HTI-601 to stain for hyaluronan (1:500). For CD31 antigen retrieval was performed using 1X Citrate Buffer pH 6 (Sigma) for 20 min, washing with 1XTBS/0.1%Tween-20 (TBST)(all washes were done in TBST), blocking with 5% goat serum for 1 hr, then incubation with primary antibody overnight at 4°C. This was followed by an endogenous peroxidase block with 3% H2O2 for 15 min, incubation with a polymer secondary antibody for 30 min (Rat Probe, BioCare Rat-on-Mouse anti-CD31 Polymer Kit), followed by Rat-HRP (BioCare) for 30 min, and then counterstaining with freshly filtered Mayer’s Hematoxylin (Fisher Scientific) for 5 min, followed by dehydration and clearing with increasing concentrations of ethanol and xylenes. For αSMA, antigen retrieval was performed using a 1X Dako Tris/EDTA pH 9 solution for 20 min. This was followed with an endogenous peroxidase block of 3% H2O2 for 10 min, blocking for avidin/biotin (Vector kit), blocking with TCT buffer (0.1% trypsin, 0.1% CaCl2, 20 mMTris-Cl pH 7.8) for 10 min, then incubation with primary antibody for 1 hour at RT. Slides were then incubated in MACH2 Rabbit HRP, followed by incubation with DAB for 10 min, counterstaining with hematoxylin, and finally dehydration and clearing. For iNos, antigen retrieval was performed using 1X Citrate Buffer pH 6 (Sigma) for 30 min, washing with 1XTBS/0.1%Tween-20 (TBST)(all washes were done in TBST), blocking with 5% goat serum for 1 hr, then incubation with primary antibody overnight at 4°C. This was followed by an endogenous peroxidase block with 3% H2O2 for 15 min, incubation with anti-rabbit HRP polymer secondary antibody for 30 min (BioCare Polymer Kit), followed incubation with DAB, and then counterstaining with freshly filtered Mayer’s Hematoxylin. To stain for HTI-601 (Halozyme): after de-waxing and rehydration, endogenous peroxide block of 0.3% H2O2 for 2 min, serum block of 2% FFA-BSA-2% normal goat serum for 30 min at room temperature, blocking with avidin/biotin (15 min each), incubation with primary antibody for 30 min at room temperature. For detection, incubate in HRP-streptavidin (BD Bioscience) for 15 min at room temperature, followed by incubation with DAB for 10 min, counterstaining with hematoxylin, then dehydration and clearing. All washes were done in TBST.

Immunofluorescent staining.

OCT frozen tissue sections were used to stain for CD11b (1:25; Abcam, ab8878), Ly6G (1:25; Bio X Cell, BP0075–1), and anti-pan cytokeratin (1:200; Millipore Sigma, F3418). Slides were air dried for 15 min at RT, incubated in acetone for 15 min, then air dried for 15 min. Slides were rehydrated with 1X PBS for 10 min, blocked with 2% normal goat serum for 1 hr at RT, and incubated with primary antibody for 2 hours at RT. Following this, secondary antibody (1:1000; Life Technologies,1749750) and directly-conjugated antibodies (PanCK) were added for 1 hour at RT, followed by counterstaining with Bisbenzimide (1:10,000; Sigma-Aldrich) for 10 min at RT, and mounting with Prolong Gold (Life Technologies). Washes between steps were done using 1XPBS. Immunfluorescence samples were imaged on a Nikon Ti-U fluorescence microscope. A subsection of FFPE tissue sections were sent to Reveal Biosciences (San Diego, CA) for IF staining, which were dual-stained for CD3/CD8, CD3/CD4, and CD4/FoxP3. For CD3/CD8 and CD3/CD4 staining, CD3 (1:100; Abcam) primary antibody was detected with Donkey anti-Rabbit IgG (H+L) secondary antibody, Alexa Fluor 546 conjugate (Life Technologies); CD8 (1:200; eBiosciences) primary antibody was detected with Goat anti-Rat IgG (H+L secondary antibody, Alexa Fluor 647 conjugate (Life Technologies); CD4 (1:200; eBiosciences) primary antibody was detected with Goat anti-Rat IgG (H+L secondary antibody, Alexa Fluor 647 conjugate (Life Technologies). Heat-induced antigen retrieval was performed with Epitope Retrieval Buffer 2 for 20 minutes. Primary antibodies were incubated overnight at 4°C. Non-specific background was blocked with 3% normal donkey serum + 3% normal goat serum in PBS-T. For CD4/FoxP3 staining, FoxP3 (1:100; Cell Signaling) primary antibody was detected with Donkey anti-Rabbit IgG (H+L secondary antibody, Alexa Fluor 546 conjugate (Life Technologies). CD4 (1:50; eBiosciences) primary antibody was directly conjugated to eFluor 660. Heat- induced antigen retrieval was performed using Leica Bond Epitope Retrieval Buffer 1 (Citrate solution, pH 6) or Epitope Retrieval Buffer 2 for 20 minutes. Primary antibodies were incubated overnight at 4°C. Non-specific background was blocked with 3% normal donkey serum in PBS-T. Whole slide fluorescent images were generated using a Pannoramic SCAN (3D Histotech). Cell staining for p53 was performed using the CM5 rabbit polyclonal antibody against p53 (1:200 Leica Biosystems; NCL-L-p53-CM5p) followed by Alexa Fluor 488 conjugate secondary antibody and Bisbenzimide.

Imaging and analysis of collagen fiber networks and small molecule distribution.

Multiphoton excitation microscopy and second harmonic generation (SHG) imaging were implemented to visualize doxorubicin distribution and fibrillar collagen in formalin-fixed paraffin embedded (FFPE) tissue sections that were rehydrated with xylenes and serial dilutions of ethanol, then mounted with Prolong Gold (Life Technologies). Visualization of collagen and doxorubicin was done on a custom-built multiphoton laser scanning microscope (Prairie Technologies/Bruker, Middleton, WI) that was described previously (30,31) using a Mai Tai Ti:Sapphire laser (SpectraPhysics, Santa Clara, CA) at an excitation wavelength of 880 nm and 800 nm, respectively.

TCGA human patient Cohort data analysis.

Transcript analysis was performed on human datasets publically available through the TDGA database. Correlation analysis was performed using the GEPIA platform (32).

Statistical analysis.

Two-group data were analyzed using a t-test. Multigroup data were analyzed using ANOVA followed by the Tukey multiple comparison post test. Percent FOV data was analyzed using Fisher’s exact test.

RESULTS

Biophysical barriers prevent effective drug delivery in PDA

As PDA progresses from preinvasive PanINs to invasive disease, an increasingly robust stromal environment develops, effectively preventing therapeutic distribution (4,9,12,33). This stromal complexity, which critically contributes to the unique pathology of PDA, is faithfully reflected in the genetically engineered KrasLSL-G12D/+;p53LSL-R172H/+;Pdx-1-Cre (KPC) mouse model (4,29,34)(Fig. 1A-T). Importantly, KPC tumors also utilize the TGF-β pathway (29), which contributes to the fibrotic and immunosuppressive desmoplastic reaction in both murine and human PDA (35,36), and activated PSCs from KPC tumors express transcripts reminiscent of fibrosis, e.g. Tgfβ1, Col1a1, and Fn1 (16). In KPC mice, α-SMA+ myofibroblasts emerge around early stage PanIN lesions and persist throughout later stages of disease (Fig 1A-C), depositing a robust ECM network (Fig. 1D-L). This includes a dense collagen matrix (Fig. 1D-I), which was confirmed to be largely fibrillar collagen from second harmonic generation (SHG) imaging (Fig. G-I), and large quantities of hyaluronan (Fig. 1J-L), a pervasive glycosaminoglycan that imbibes fluid (7,8), contributing to grossly elevated interstitial fluid pressure (IFP) in PDA (Fig. 1T), consistent with previous findings (4). When IFP increases to these levels, convective transvascular transport (i.e. intravascular to interstitial pressure-driven flux) is significantly hindered, and therapeutics must now be transported without, or even against, a pressure gradient. Moreover, compounding the unique hypovascularity of PDA (3,4)(Fig. 1S), many blood vessels actually collapse under the pressure (4), leaving a notable paucity of functional vessels in PDA, and to some degree in preinvasive disease (Fig. 1M-O). Indeed, CD31 immunohistochemistry (IHC) reveals a stark difference in blood vessel patency phenotypes between healthy pancreata with abundant normal vessels with apparent lumens, and pancreas tumors with a predominantly compressed vessel phenotype (i.e. no visible lumen)(Fig. 1M-O). Previously, we and others have shown that these phenotypes correlate with either fully or non-perfused vasculature, respectively (3,4,9). The collective impact of these stromal barriers on drug delivery can be visualized by analyzing doxorubicin distribution in normal pancreata, preinvasive, and invasive disease (Fig. 1P-R). A clear reduction in doxorubicin distribution and nuclear uptake is evident in diseased pancreata, and is increasingly pronounced with disease progression (Fig. 1Q,R).

Figure 1. Desmoplasia creates stromal barriers and limits drug delivery in PDA.

Figure 1.

(A-C) Activated PSCs express α-SMA+, emerge during preinvasive disease, and remain prevalent throughout PDA. (D-F) Masson’s trichrome staining shows collagen (blue) deposition by myofibroblasts during disease. (G-I) Fibrillar collagen determined from second harmonic generation imaging. (J-L) Intense hyaluronan expression during disease. (M-O) CD31 immunohistochemistry shows transition from open vessels in normal pancreata (arrowheads) to increasingly showing the phenotype of vascular collapse (arrows). (P-R) Decreased doxorubicin distribution in disease (after i.v. injection 15–20 minutes before euthanasia). S) PDA is hypovascular (n>6 regions/condition). (T) Extremely elevated interstitial fluid pressure (IFP) in PDA (n=4/group). (U) Key ECM and TGF-β pathway transcripts (n=178 TCGA patients) are upregulated in human PDA. (V) Strong correlation between α-SMA (ACTA2) and collagen-I (COL1A1) expression in human PDA. (W) Collagen expression relates to survival in human patient (TCGA PDA dataset). (X-Z) Human TCGA data shows Strong correlation between genetic collagen-I expression and key profibrotic TGF-β signaling cascade components. (Scale bar=25μm, p****<0.0001, Data are mean±SEM)

Analysis of transcript data from the TCGA human PDA patient cohort likewise demonstrates robust expression of stromal ECM transcripts (Fig. 1U), as expected, e.g. (37,38). Further analysis reveals a significant correlation between α-SMA expression and expression of fibrotic ECMs, including type-I collagen, fibronectin, and hyaluronan synthases (Fig. 1V; Supplementary Fig. S1), with the strongest correlation between COL1A1 and α-SMA (ACTA2)(Supplementary Fig. S1). We note that high expression of type-I collagen, present in the majority of PDA patients, results in poorer survival outcome compared to the patient population with low expression (Fig. 1W). In fact, the 2-year survival for patients with low collagen expression levels is relatively high and surprisingly stable, suggesting that COL1A1 may be a useful biomarker to predict outcome, and that reducing collagen levels in PDA may be beneficial.

To better understand signal transduction associated with fibrosis, we next evaluated relationships between TGF-β signaling elements, which are elevated in human PDA compared to normal pancreata (Fig. 1U), and stromal ECM. Analysis demonstrates significant relationships between TGF-β pathway (i.e. SMAD2, SMAD3, TGFB1, and TGFB2) and stromal ECM transcripts (COL1A1, COL1A2, and HAS1, 2, and 3), with the strongest correlations for collagen (Figs. 1X-Z; Supplementary Fig S1). Similar relationships were observed between transcripts for type-I collagen or hyaluronan synthases and TGF-β receptors transcripts TGFBR1 or TGFBR2. Thus, these combined data in human and murine pancreas cancers suggest that targeting activated PSCs and collagen through the TGF-β pathway is a viable strategy to combat stromal fibrosis in PDA.

Halofuginone is a potent antifibrotic agent in autochthonous PDA

The potent antifibrotic efficacy of halofuginone (HF) have been demonstrated in pancreatitis and when administered near the onset of subcutaneously grafted pancreatic tumors (24,25). However, HF has yet to be evaluated in autochthonous PDA, particularly during intervention trials. First, to study of HF in vitro, primary PSCs derived from both human and KPC autochthonous tumors (Supplementary Fig. S2A-H) were treated with 0, 50, and 100 nM HF for 48 hours, demonstrating a significant, dose-dependent inhibition of per-cell α-SMA expression (i.e. PSC activation) in both mouse and human cells (Fig. 2A-C). Not surprisingly, inhibition of α-SMA expression corresponds to reduced expression of fibrosis-related genes, such as type-I collagen (Col1a1) and hyaluronan synthase 2 (Has2)(Fig. 2D), and consistent with reports targeting PSCs with Losartan (9,10). Note, since Has2 is expressed by activated PSCs at ~17X and ~85X the level of Has1 and Has3, respectively Supplementary Fig. S2I), we focused our analysis on Has2. Furthermore, both human and murine PSCs exhibit decreased proliferation at higher HF doses (Fig. 2E).

Figure 2. Halofuginone inhibits fibrotic activity in vitro and in vivo.

Figure 2.

(A-C) Immunofluorescent staining of DAPI (blue) and α-SMA (green) in murine (B) and human (C) PSCs shows dose-dependent inhibition of α-SMA (scale bar=10μm; n=3/condition with 5 ROIs analyzed/sample). (D) Expression (via qPCR) of the fibrosis-related genes Col1a1 (collagen-I) and Has2 (hyaluronan synthase-2) are significantly inhibited with increasing HF concentration. (E) Dose-dependent proliferation inhibition (MTS proliferation assay) in murine (mPSC) and human PSCs (hPSC) following 48 hours of HF treatment. (F,G,N) In vitro observations are maintained in vivo, where α-SMA immunohistochemical reveals a decrease in PSC activation (α-SMA+) with HF. (H-Q) Consistent with these results, there was a Significant decrease in hyaluronan (H,I,O) and collagen (via Trichrome staining (J,K,P) and SHG for fibrillar collagen (L,M,Q)). (n=6 control; n=5 HF-treated mice;10 ROIs analyzed/tumor) (Scale bar=50 μm, p*<0.05, p**<0.01, p***<0.001, p****<0.0001, Data are mean±SEM)

Compellingly, the antifibrotic effects of HF seen in vitro were maintained in vivo in physiologically relevant tumors. KPC mice were monitored and enrolled in 10-day intervention studies once tumors reached a diameter of 4–9mm in largest dimension, as evaluated using high-resolution ultrasound (Fig. 2F-M). Consistent with the results observed in vitro, mice treated with HF exhibited a significant decrease in stromal α-SMA+ signal, indicating a decrease in density of activated PSCs (Fig. 2F,G,N) and a profound effect on the stromal ECM. HF-treated animals showed a significant decrease in hyaluronan (Fig. 2H,I,O) and total collagen content, as determined through a Masson’s trichrome stain (Fig. 2J,K,P), with SHG imaging confirming this effect in fibrillar collagen (Fig. 2L,M,Q), underscoring the utility of HF as a potent antifibrotic agent in PDA.

Antifibrotic therapy facilitates drug delivery in established PDA

Hypertension is a characteristic property of virtually all solid tumors, disrupting natural pressure gradients and dramatically reducing convective transport (7,8,39). As a result, diffusive transport through the viscous, charged, tortuous, and sterically-challenging interstitial environments becomes the primary mechanism driving therapeutic distribution (8,39). We note that collagen content has been identified as a main determinant of interstitial transport, with hyaluronan also limiting transport by absorbing residual fluid, resulting in increased interstitial viscosity and intratumoral pressure (7,39). Therefore, we evaluated the impact of HF-induced depletion of collagen and hyaluronan on molecular transport in PDA. We found that stromal re-engineering to normalize tumor microenvironments with antifibrotic therapy markedly disrupts transport barriers associated with PDA (Fig. 3). IFP levels are significantly reduced in HF-treated animals (Fig. 3A), substantially increasing transvascular flux of small molecule therapy. Furthermore, relieving tumor pressure allows previously collapsed, nonfunctional vessels to open, leading to a significant increase in the percent of functional, perfused, vessels (Fig. 3B-F) without an increase in vessel density consistent with findings following PEGPH20 therapy but distinct from increased vessel density following Hh treatment (3,4). As a result of disrupting biophysical barriers to transport in PDA, a significant improvement in small molecule distribution is observed (Fig. 3G-O, Supplementary Fig. S3A,B), where doxorubicin distribution (Fig. 3P) negatively correlates with fibrotic area (Fig. 3Q). Thus, these data suggest that antifibrotic therapy with HF effectively disrupts barriers to therapeutic delivery and distribution, which is increasingly needed to enhance delivery of peptide and antibody therapies (e.g. molecular and immune therapies), and thus, may represent an effective component of carcinoma-stromal-immune combination therapy strategies.

Figure 3. Antifibrotic therapy facilitates drug delivery in autochthonous PDA.

Figure 3.

(A) Decreased IFP following antifibrotic therapy (n=4 WT; n=4 Control KPC, n=3 HF-treated KPC). (B,C) CD31 immunohistochemistry from control and HF-treated KPC tumors. (D,E) Vessel perfusion with FITC-conjugated dextrans (100μL of 10mg/mL solution injected intracardiac 5 minutes prior to euthanasia). (F) Improved vessel perfusion following HF treatment (perfused vessels/total vessel; n=3 mice/group; ≥5 ROI assessed/tumor). (G-I) Clear recovery of doxorubicin distribution (following i.v. injection 15–20min before euthanasia) in the HF-treated tumors (I), in contrast to little to no perfusion in control tumors (H). (J-L) Significantly reduced fibrillar collagen density and organization (via SHG) (M-O) Merging doxorubicin and SHG data illustrates improved drug distribution correlating with lower collagen density. (P,Q) Quantification of doxorubicin distribution (P) and collagen (Q)(n=3 mice/group; 5 ROI assessed/tumor). Scale bar=50 μm, p*<0.05, p**<0.01, p***<0.001, p****<0.0001, Data are mean±SEM.

Halofuginone performs as a dual-action therapy by targeting pancreatic carcinoma cells

HF is capable of inhibiting PSC activation and collagen production in vitro, even at very low, non-terminal levels (Fig. 2). At higher concentrations, PSCs exhibit dose-dependent inhibition of proliferation (Fig. 2). Interestingly, we also observe decreased proliferation of primary pancreatic carcinoma cells in vitro following HF treatment (Fig. 4A). This indicates the possibility of a dual-action role for HF as a PDA therapy in which carcinoma cells are impacted in addition to stroma-targeting effects. A number of targeted therapies have been reported to enhance carcinoma cell sensitivity to chemotherapy (4042), including targeting CTGF, a member of the TGF-β signaling family (40). This, in combination with our proliferation data, led us to question whether HF might increase susceptibility of carcinoma cells to gemcitabine (Gem), a standard-of-care chemotherapeutic used to treat pancreatic cancer. To test this hypothesis, cells were subjected to various combination doses of HF and Gem, selected based on dose-response curves we established for each agent (Fig. 4A,B). Equal numbers of cells were allowed to adhere for 24 hours before HF-supplemented media was added for 24 hours. HF-supplemented media was then replaced by Gem-supplemented media for an additional 24 hours. Finally, supplemented media was removed and replaced with normal media for 2 hours, at which point a MTS cell proliferation assay assessed cell viability (Fig. 4C,D). Interestingly, the relative decrease in proliferation with increasing gemcitabine concentration remains fairly static (~66%), regardless of HF concentration used to pre-treat the cells, revealing that pre-treatment of carcinoma cells with HF does not increase sensitivity to gemcitabine (Fig. 4D). It is, however, important to note that gemcitabine treatment following HF therapy can further reduce the carcinoma cell population, suggesting that while HF may be an effective monotherapy, greater cell death may be achieved with HF in concert with chemo and/or immune therapies.

Figure 4. Halofuginone inhibits proliferation of carcinoma cells without enhancing chemotherapy.

Figure 4.

(A,B) Dose-response curve of HF-treated (A) or gemcitabine-treated (B) primary murine carcinoma cells (mPDA). (C) Proliferation assay timeline to test ability of HF to sensitize cells to gemcitabine. (D) Results of Sensitization assay showing that the relative response to gemcitabine remains largely static (~66% decrease in proliferation), regardless of HF pre-treatment concentration, indicating that HF does not increase sensitivity of carcinoma cells to gemcitabine (n=3 replicates/condition).

Antifibrotic therapy promotes influx and distribution of macrophages and cytotoxic T cells in PDA

Although immunotherapies are showing great promise in a number of cancers, their impact on PDA has remained modest. This is due, in part, to low neoantigen load, strongly immunosuppressive microenvironments, and a dense stroma, which can physically limit both drug and cell infiltration (2,5,6,4345). However, studies that targeted the stroma in concert with checkpoint inhibition have observed strong immune response in PDA (16,45), demonstrating that a robust anti-tumor immune response can take place in PDA under the right therapeutic conditions. Thus, stromal re-engineering to normalize tumor microenvironments may not only facilitate delivery of therapeutics (to combat disease and aid antigen presentation), but also to promote distribution of immune therapies (e.g. checkpoint inhibition, cytokine blockade, etc.) and anti-tumor immune cell populations. Yet, our understanding of immune dynamics and surveillance following stroma targeting is currently limited. Therefore, to determine if HF results in immunomodulatory effects, we examined tissue from mice 24hr and 10-day after initiation of HF intervention therapy. Examination of H&E tumor sections suggested that mice treated with HF for only 24 hours exhibit a striking increase in immune infiltrate, an effect that appears to subside in the 10-day group (Fig. 5A-C). To quantify histopathology observations of immune infiltrate in H&E sections, we created an image-processing algorithm to distinguish and count immune cells based upon nuclear characteristics. The algorithm selected for nuclei that were smaller than the comparably larger nuclei of carcinoma cells, but larger than random debris, with circularity set to 0.75–1.00 to filter out the elongated nuclei of fibroblasts (Supplementary Fig. S4). During development, the validity of this algorithm was confirmed in a subset of slides stained for CD45. This analysis confirmed a statistically significant increase in immune infiltrate 24 hours after HF treatment that appears to normalize, in terms of cell numbers, by 10 days (Fig. 5D), demonstrating that HF treatment can alter the immune landscape in PDA. Despite this compelling result, this metric did not tell us which immune cell subpopulations were being altered, differences in immune composition between groups, or whether the composition in the 10-day group was altered. We therefore performed a more focused examination of distinct immune cell populations that have been implicated as key regulators of the immune response in PDA.

Figure 5. Antifibrotic therapy modulates the immune landscape.

Figure 5.

(A-C) Histopathology analysis of KPC tumor H&E sections suggests a dramatic increase in immune infiltrate 24hr after HF treatment (blowup panels: arrows highlight immune cells; arrowheads highlight larger carcinoma cell nuclei associated). (D) Quantification of immune infiltrate in H&E sections confirming a significant influx of immune cells 24hrs after HF treatment (determined using an algorithm to detect histologic immune infiltrate by filtering nuclei on the basis of size, shape, and density; n=3 mice/group; ≥3 ROI/tumor). (E) Significant Cd11b+ cell increases after 24hrs and 10d revealing that the majority of immune infiltrate is myeloid cells. (F) Quantification of Ly6G+ cells, a validated marker for immunosuppressive CD11b+ MDSCs in PDA, showed no significant difference between treatment groups. (G) Quantification of iNos+ cells, a functional marker for classically activated macrophages with inflammatory functions, shows significant increases after HF treatment. (H, I) No significant difference in (H) CD3+;CD4+ T helper cells or (I) CD4+;FoxP3+ Treg cell populations. (J) Quantification of CD3+;CD8+ dual IF showing a significant increase in cytotoxic CD8+ T cells in tumors treated for 24 hours. (K) Percent FOVs with at least one CD8+ T cell, showing a significant increase in the distribution of CD8+ T cells throughout PDA tumors. For E-K n≥5 control, n=3 24hr, n=5 10d mice with ≥4 ROI/tumor. Scale bar = 50 μm, p*<0.05, p****<0.0001, Data are plotted as mean±SEM.

Inspection of specific immune populations (Fig. 5E-J) through immunofluorescence (IF) and IHC staining of KPC tumors (Supplementary Fig. S5A-R) revealed a significant increase in CD11b+ myeloid cells (Fig. 5E) and CD8+ cytotoxic T cells (Fig. 5J,K). Because CD11b is a broad myeloid cell marker, expressed on both protumorigenic (e.g. myeloid derived suppressor cells (MDSCs), “M2” alternatively activated macrophages) and anti-tumorigenic (e.g. “M1” classically activated macrophages) cells, we sought to further define this population. Analysis of IF staining for Ly6G, a validated marker for a primary immunosuppressive population of CD11b+ MDSCs in PDA (5), shows no significant difference in the number of Ly6G+ cells between groups (Fig 5F). However, there is an insignificant trend suggesting a decrease in Ly6G+ cells in both HF treatment groups, indicating that the increase in CD11b+ cells cannot be attributed to increased infiltration of immunosuppressive MDSCs. In contrast, analysis of IHC staining for iNOS, a functional marker of classically activated inflammatory macrophages, shows significant increase in the presence and distribution of iNOS+ cells following HF treatment (Fig. 5G, Supplementary Fig. 5G-I). The iNOS data is consistent with the trends observed in the Cd11b+ data, suggesting that inflammatory macrophages are a chief contributor to the immune influx observed at 24 hours and remain significantly elevated at 10 days, demonstrating a shift in the immune composition during HF treatment. Furthermore, analysis of T cell populations showed no significant difference for T helper cells (CD3+CD4+) or T regulatory cells (CD4+Foxp3+) (Fig. 5H,I), but reveals a significant increase in CD8+ cytotoxic T cells (Fig. 5J). This is particularly compelling when considered in the context of overall intratumoral distribution. Over half of the fields of view (FOV) analyzed in untreated tumors show a complete absence of CD8+ cells (Fig 5J), which may be related to reports that the robust ECM in PDA may impede T cell infiltration or distribution (46,47). In stark contrast, the 24hr HF group was completely negative for CD8+ cells in only 23% of FOVs (Fig. 5J), indicating a robust increase in the distribution of CD8+ T cells. These data show that targeting the stroma has the capacity to increases numbers, distribution, and thus sampling, of CD8+ T cells, which are capable of killing multiple cancer cells in a region. Thus, although the mere presence of intratumoral T cells does not guarantee an anti-tumor response, the ability to increase CD8+ cell infiltration in PDA is an indicator of overcoming two key immunosuppressive pathways in PDA, i.e. physical exclusion by ECM barriers and chemical exclusion via cytokines (16,45,47). Therefore, these data suggest that stromal re-engineering approaches can generate an enhanced anti-tumor immune response, but may require additional immune modulating therapy, including checkpoint inhibition, to enhance and maintain an anti-tumor immune response.

Regions of decreased hyaluronan are associated with increased immune infiltrate

To better understand the stromal changes that influence immune infiltrate into PDA, we revisited our investigation of the ECM (Fig. 6A-I). SHG imaging ruled out fibrillar collagen content as an explanation, since there is no significant difference between the 24hr and control groups (Fig. 6G-J). However, interestingly, we observed a “relaxation” of the collagen matrix, made evident by increased fiber waviness (i.e. greater crimp under low stress conditions that likely produce the trend of modestly increasing area fraction; Fig. 6H,J)). This is consistent with decreased myofibroblast behavior and decreased hyaluronan swelling pressures that are constrained by collagen networks (4,8), suggesting that the more relaxed collagen matrix may pose less of a barrier to immune cell infiltration and that changes in the hyaluronan matrix may be occurring. Indeed, we discovered a profound relationship between overall and regional hyaluronan levels and immune infiltrate (using nuclear phenotype algorithm analysis and visual confirmation in adjacent H&E sections) (Fig. 6A-F). In control mice, analysis of hyaluronan levels and regions of immune infiltration show an inverse correlation, with greater infiltrate associated with lower hylauronan (Fig. 6K,L). Moreover, we observed, perhaps surprisingly, that rapid changes in hylauronan emerge, with more regions containing low levels of hyaluronan just 24hr after HF treatment (Fig. 6D-F and 6J), suggesting that some degradation of hyaluronan has occurred. Interestingly, in the 24h group the relationship between hyaluronan levels and immune infiltration holds, and therefore greater frequency of hyaluronan-low areas in 24hr-group tumors are associated with a greater immune cell numbers, showing that decreased hyaluronan facilitates greater immune infiltration into PDA. Indeed, comparison of immune cells in “low HA” and “high HA” conditions (bottom and top 50% of signal for each group, respectively) demonstrates that there are greater numbers of immune cells in HA-low regions compared to HA-high regions for both the control and HF-treated mice, and significant increases in immune cells due to HF treatment (Fig. 6M), consistent with the increase in HA-low regions following HF treatment (Fig. 6E,K). Thus, we conclude that HF treatment results in a rapid impact on HA composition and, thereby, collagen matrix architecture (i.e. matrix loosening likely from decreased hyaluronan-generated swelling pressure), promoting immune infiltration into PDA.

Figure 6. Immune infiltrate in PDA correlates with regions of decreased Hyaluronan.

Figure 6.

(A-C) H&E staining of untreated (A) and HF-treated (B,C) KPC tumors and adjacent sections stained for hyaluronan (D-F), showing regions of histologically-identifiable immune infiltrate have lower hyaluronan. In addition, following HF treatment hyaluronan-low regions are larger and more frequent (Asterisk: example of lower HA regions that are immune cell rich). (G-I) SHG analysis shows a collagen matrix loosening (i.e. increased fiber waviness, or crimp, associated with low stress conditions) 24hr after HF treatment and decreased collagen at 10d. (J) Collagen fraction analysis from SHG imaging showing a slight increase 24h after HF treatment, consistent with increased waviness, and a significant decrease after 10 days. (K) Significant decreases in hyaluronan following HF treatment (n≥3/condition with ≥5 ROI assessed per tumor). (L) Significant, moderate, correlation (p<0.006; r=0.−46) between hyaluronan (HA) area fraction and immune cell numbers showing increasing immune cell number with lower HA. (M) Comparison of immune cells in low and high HA regions (bottom and top 50% of signal for each group, respectively) showing greater immune cell numbers in low vs. high regions, and significantly increased immune infiltrate cells after HF treatment, consistent with increased low HA regions following HF treatment. (p*<0.05, p**<0.01, p***<0.001. Scale bar A-F = 100 μm; Scale bar G-I = 50 μm; data are plotted as mean±SEM)

Halofuginone treatment leads to widespread necrosis and reduced tumor volume growth

While HF alters immune dynamics and is an effective inhibitor of fibrosis and proliferation, macroscopic changes in tumor morphology speak to a more global, clinically-relevant outcome. To assess such changes, we measured tumor volume growth via high-resolution ultrasound in KPC mice enrolled in the 10-day intervention group (Fig. 7A-D). HF-treated mice exhibit significantly lower tumor volume growth compared to control mice, with one tumor decreasing in volume (Fig. 7E). Notably, and consistent with our quantitative measures of tumor volume, widespread necrosis is present in tumors from mice treated with HF for 10 days (Fig. 7F-H). Indeed, over 50% of the treated mice show necrosis in >20% of the tumor area, with one extreme responder displaying necrosis in over 50% of the evaluable tumor (Fig. 7H), with no observable toxicity or necrosis to adjacent normal pancreas (Supp. Fig 6A,B) or significant differences in body weight (Supp. Fig 6C). Overall, these data suggest that HF monotherapy significantly impacts PDA, but may be particularly useful in concert with single agent or combinations of molecular and/or immune therapies. Thus, we conclude that HF treatment results in reduced PSC activation inducing alterations in ECM levels and architecture and decreased carcinoma cell and CAF viability; this facilitates infiltration of anti-tumor immune populations, and in particular macrophages that may influence necrosis and help orchestrate T cell responses, and culminates with large regions of necrosis and reduced tumor growth. Therefore, while a series of reductionist and cell-depletion studies are required to parse out the specific impact of each cell population, data presented here clearly demonstrate that HF has strong potential to both improve drug delivery and promote infiltration of anti-tumor immune populations, warranting further study as part of a combination molecular and immune therapy approach to combat metastatic PDA.

Figure 7. Halofuginone induces widespread necrosis and inhibits tumor volume growth.

Figure 7.

(A-D) 3D reconstructed tumor volumes in control versus HF-treated KPC mice. (E) Quantification of the relative change in 3D tumor volume following one week of treatment (n=5 control; n=7 HF). (F,G) Intratumoral necrosis in a control versus HF-treated KPC tumor after 10 days (green=PDA region, red=necrotic region. (H) Quantification of percent necrosis observed within the defined PDA tumor region at 10-day study endpoint (n=9 Control; n=6 HF). Scale bar = 2 mm, p*<0.05, data are mean±SEM.

DISCUSSION

HF appears to be a valuable therapeutic agent, disrupting barriers to drug delivery and inducing infiltration of anti-tumor immune populations. It improves drug transport into and through tumors by enhancing convection and diffusion, which is consistent with, and provides support for, previous conclusions that inordinately high IFP in PDA results from HA-driven swelling pressures constrained by a collagen network (4,8). Furthermore, our analysis demonstrates that the small subpopulation of patients with low collagen expression show better survival outcome, consistent with recent IHC analysis (48) and suggesting that reducing collagen in PDA may be generally beneficial. Furthermore, the marked immune response and shift in immune composition after HF administration, suggests that HF may be applied synergistically to enhance immune therapy approaches, such as immune checkpoint inhibitors or agonists for tumoricidal macrophages. Indeed our findings appear consistent with recent work demonstrating that FAK hyperactivity in PDA promotes stromal collagen and plays a role in immunosuppression, partially explaining the failure of immunotherapy in PDA while also demonstrating enhanced anti-tumor immunity with stromal disruption and checkpoint therapy (45). Lastly, we note that while HF impacted the primary tumor, equally impactful measures are effects on metastases. Unfortunately, a limited number and heterogeneity of evaluable macrometastases in distinct organ sites (n=3 HF; n=4 Control) restricted statistically powerful conclusions. However, we note that one HF-treated mouse exhibited almost complete metastasis necrosis. Likewise, a pilot case study on one mouse treated over >6.5 months showed large, but almost entirely necrotic lesions in the liver. Thus, our data suggest that further mechanistic studies and full preclinical trials with HF in combination with chemo and immune therapies are warranted.

In light of our findings, and reports implementing diverse stroma-targeting approaches (3,4,10,16,18,19,45), it is clear that stromal re-engineering to normalize tumor microenvironments holds promise to improve PDA outcomes, particularly when combined with effective cytotoxic agents. However, not all stroma-targeting approaches have demonstrated clinical benefit to date. For instance, following groundbreaking work defining hypovascularity and the potential for Hh inhibition to alter the stroma and improve chemotherapy delivery in autochthonous PDA (3), several clinical trials were initiated, eventually resulting in conflicting findings using either IPI-926 or vismodegib (20,49,50). One Phase-Ib/II trial (IPI-926+gemcitabine; NCT01130142) was terminated early when data suggested adverse effects, while a separate trial (IPI-926+FOLFIRINOX; NCT01383538 (20)) suggested therapeutic benefit, consistent with recent reports targeting Hh in preclinical models (18,19). In an attempt to understand these findings, recent work utilized genetic disruption or prolonged pharmacologic inhibition of Hh starting from early preinvasive disease, and showed that resultant disease appears more aggressive, has increased vascularity, and results in decreased survival (17,51,52). A different approach to define and combat stromal barriers in late-stage PDA, demonstrated that ablating stromal hyaluronan with PEGPH20 in the context of concomitant chemotherapy normalizes tumor pressures with concurrent re-expansion of collapsed microvasculature and a dramatic increase in drug delivery, leading to reduced metastasis and improved survival(4). This is consistent with the data here, highlighting the utility of targeting a physical property of the tumor and its clinical impact. Indeed, a multi-therapeutic PEGPH20 regimen is currently in phase-III trial (NCT02715804; with multiple phase-II trials ongoing), following phase-II data demonstrating a significant benefit in HA-high PDA patients(53). Likewise, angiotensin inhibition reduces collagen production in CAFs through TGF-β1 inhibition and has shown great promise in grafted tumors(9,10). Although this approach has yet to be directly tested against autochthonous PDA, it suggests mechanistic overlap with HF and bolsters the case for antifibrotic agents against established PDA. Thus, collectively, while these findings make it clear that great care must be taken when developing rational strategies to perturb CAFs and/or normalize the stroma, they also demonstrate a need to increase our understanding of both physical and molecular mechanisms regulating stromal dynamics in order to develop effective stroma-targeting strategies. Indeed, some of the disparities between the studies may result from a protective role for fibroblasts in early PDA (17), in agreement with early reports suggesting that normal fibroblasts may help prevent or limit disease (54), and recent findings of CAF heterogeneity in PDA (15). It is also plausible that our current understanding of Hh signaling across multiple cell populations associated with PDA tumors is incomplete, particularly since Hh inhibition studies show an angiogenic response that has not been observed with other stroma-targeting approaches. Thus, the implications of targeting Hh signaling may extend beyond suppression of paracrine signaling between carcinoma cells and myofibroblasts. Overall, results suggest that different stages of PDA stages may respond quite differently to distinct stroma-targeting approaches and that certain stroma-targeting approaches may present a specific therapeutic window where the tumor is susceptible to subsequent therapy, and that it may be unwise to administer some agents long-term. Hence, we clearly need to increase our understanding of the physical and molecular stromal dynamics to develop sound stroma-targeting strategies that will likely be an instrumental component to treating PDA.

Supplementary Material

1

STATEMENT OF SIGNIFICANCE.

This work demonstrates how focused stromal re-engineering approaches to normalize the stroma disrupt physical barriers to effective drug delivery and promote anti-tumor immunity.

ACKNOWLEDGEMENTS

P.P. Provenzano and this work was supported by the NIH (R01CA181385 and U54CA210190 University of Minnesota Physical Sciences in Oncology Center to P.P. Provenzano, and a SPORE Development award under P50CA101955 to P.P. Provenzano) and by a Research Scholar Grant, RSG-14–171-01-CSM from the American Cancer Society (to P.P. Provenzano). This work was also supported by the Randy Shaver Research and Community Fund, UMN College of Science and Engineering, Masonic Cancer Center, and grants from the UMN Institute for Engineering in Medicine (all to P.P. Provenzano). K. Elahi-Gedwillo received support from the Dept. of Education (GAANN fellowship through grant #P200A120198) and ARCS (Achievement Rewards for College Scientists Foundation, Inc.). The content of this work is solely the responsibility of the authors and does not necessarily represent the official views of the NIH or other funding agencies. The authors thank Courtney Podritz and Ben Cooper for their assistance with pilot experiments. We would also like to thank Dr. Ajay Dixit for isolating the human-derived primary PSCs used in our in vitro studies and members of the Provenzano laboratory for insightful comments regarding this work.

Footnotes

CONFLICTS OF INTEREST

There are no conflicts of interest to declare.

REFERENCES

  • 1.American Cancer Society Cancer Facts and Figures 2018:18. [Google Scholar]
  • 2.Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer research. 2007;67:9518–27. [DOI] [PubMed] [Google Scholar]
  • 3.Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer cell. 2012;21:418–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stromnes IM, Brockenbrough JS, Izeradjene K, Carlson MA, Cuevas C, Simmons RM, et al. Targeted depletion of an MDSC subset unmasks pancreatic ductal adenocarcinoma to adaptive immunity. Gut. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stromnes IM, Hulbert A, Pierce RH, Greenberg PD, Hingorani SR. T-cell Localization, Activation, and Clonal Expansion in Human Pancreatic Ductal Adenocarcinoma. Cancer immunology research. 2017;5:978–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.DuFort CC, DelGiorno KE, Carlson MA, Osgood RJ, Zhao C, Huang Z, et al. Interstitial Pressure in Pancreatic Ductal Adenocarcinoma Is Dominated by a Gel-Fluid Phase. Biophysical journal. 2016;110:2106–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Provenzano PP, Hingorani SR. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. British journal of cancer. 2013;108:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV, et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nature communications. 2013;4:2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:2909–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson JS, et al. Pancreatic stellate cells and pancreatic cancer cells: an unholy alliance. Cancer research. 2008;68:7707–10. [DOI] [PubMed] [Google Scholar]
  • 12.Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, Tuveson DA. The pancreas cancer microenvironment. Clinical cancer research : an official journal of the American Association for Cancer Research. 2012;18:4266–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer research. 2008;68:918–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Xu Z, Vonlaufen A, Phillips PA, Fiala-Beer E, Zhang X, Yang L, et al. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am J Pathol. 2010;177:2585–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. The Journal of experimental medicine. 2017;214:579–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:20212–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer cell. 2014;25:735–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mpekris F, Papageorgis P, Polydorou C, Voutouri C, Kalli M, Pirentis AP, et al. Sonic-hedgehog pathway inhibition normalizes desmoplastic tumor microenvironment to improve chemo- and nanotherapy. Journal of controlled release : official journal of the Controlled Release Society. 2017;261:105–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zhao J, Wang H, Hsiao CH, Chow DS, Koay EJ, Kang Y, et al. Simultaneous inhibition of hedgehog signaling and tumor proliferation remodels stroma and enhances pancreatic cancer therapy. Biomaterials. 2018;159:215–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ko AH, LoConte N, Tempero MA, Walker EJ, Kate Kelley R, Lewis S, et al. A Phase I Study of FOLFIRINOX Plus IPI-926, a Hedgehog Pathway Inhibitor, for Advanced Pancreatic Adenocarcinoma. Pancreas. 2016;45:370–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nelson EF, Huang CW, Ewel JM, Chang AA, Yuan C. Halofuginone down-regulates Smad3 expression and inhibits the TGFbeta-induced expression of fibrotic markers in human corneal fibroblasts. Molecular vision. 2012;18:479–87. [PMC free article] [PubMed] [Google Scholar]
  • 22.Keller TL, Zocco D, Sundrud MS, Hendrick M, Edenius M, Yum J, et al. Halofuginone and other febrifugine derivatives inhibit prolyl-tRNA synthetase. Nature chemical biology. 2012;8:311–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Spector I, Honig H, Kawada N, Nagler A, Genin O, Pines M. Inhibition of pancreatic stellate cell activation by halofuginone prevents pancreatic xenograft tumor development. Pancreas. 2010;39:1008–15. [DOI] [PubMed] [Google Scholar]
  • 24.Spector I, Zilberstein Y, Lavy A, Nagler A, Genin O, Pines M. Involvement of host stroma cells and tissue fibrosis in pancreatic tumor development in transgenic mice. PloS one. 2012;7:e41833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zion O, Genin O, Kawada N, Yoshizato K, Roffe S, Nagler A, et al. Inhibition of transforming growth factor beta signaling by halofuginone as a modality for pancreas fibrosis prevention. Pancreas. 2009;38:427–35. [DOI] [PubMed] [Google Scholar]
  • 26.Bruck R, Genina O, Aeed H, Alexiev R, Nagler A, Avni Y, et al. Halofuginone to prevent and treat thioacetamide-induced liver fibrosis in rats. Hepatology. 2001;33:379–86. [DOI] [PubMed] [Google Scholar]
  • 27.de Jonge MJ, Dumez H, Verweij J, Yarkoni S, Snyder D, Lacombe D, et al. Phase I and pharmacokinetic study of halofuginone, an oral quinazolinone derivative in patients with advanced solid tumours. European journal of cancer. 2006;42:1768–74. [DOI] [PubMed] [Google Scholar]
  • 28.Ruckert F, Aust D, Bohme I, Werner K, Brandt A, Diamandis EP, et al. Five primary human pancreatic adenocarcinoma cell lines established by the outgrowth method. The Journal of surgical research. 2012;172:29–39. [DOI] [PubMed] [Google Scholar]
  • 29.Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer cell. 2005;7:469–83. [DOI] [PubMed] [Google Scholar]
  • 30.Ray A, Lee O, Win Z, Edwards RM, Alford PW, Kim DH, et al. Anisotropic forces from spatially constrained focal adhesions mediate contact guidance directed cell migration. Nature communications. 2017;8:14923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Carlson M, Watson AL, Anderson L, Largaespada DA, Provenzano PP. Multiphoton fluorescence lifetime imaging of chemotherapy distribution in solid tumors. J Biomed Opt. 2017;22:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic acids research. 2017;45:W98–W102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Neesse A, Algul H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64:1476–84. [DOI] [PubMed] [Google Scholar]
  • 34.Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer cell. 2003;4:437–50. [DOI] [PubMed] [Google Scholar]
  • 35.Principe DR, DeCant B, Mascarinas E, Wayne EA, Diaz AM, Akagi N, et al. TGFbeta Signaling in the Pancreatic Tumor Microenvironment Promotes Fibrosis and Immune Evasion to Facilitate Tumorigenesis. Cancer research. 2016;76:2525–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yen TW, Aardal NP, Bronner MP, Thorning DR, Savard CE, Lee SP, et al. Myofibroblasts are responsible for the desmoplastic reaction surrounding human pancreatic carcinomas. Surgery. 2002;131:129–34. [DOI] [PubMed] [Google Scholar]
  • 37.Gress TM, Muller-Pillasch F, Lerch MM, Friess H, Buchler M, Adler G. Expression and in-situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. International journal of cancer Journal international du cancer. 1995;62:407–13. [DOI] [PubMed] [Google Scholar]
  • 38.Gress TM, Menke A, Bachem M, Muller-Pillasch F, Ellenrieder V, Weidenbach H, et al. Role of extracellular matrix in pancreatic diseases. Digestion. 1998;59:625–37. [DOI] [PubMed] [Google Scholar]
  • 39.Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nature reviews Clinical oncology. 2010;7:653–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Neesse A, Frese KK, Bapiro TE, Nakagawa T, Sternlicht MD, Seeley TW, et al. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:12325–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Muerkoster S, Arlt A, Witt M, Gehrz A, Haye S, March C, et al. Usage of the NF-kappaB inhibitor sulfasalazine as sensitizing agent in combined chemotherapy of pancreatic cancer. International journal of cancer Journal international du cancer. 2003;104:469–76. [DOI] [PubMed] [Google Scholar]
  • 42.Ohmori T, Yang JL, Price JO, Arteaga CL. Blockade of tumor cell transforming growth factor-betas enhances cell cycle progression and sensitizes human breast carcinoma cells to cytotoxic chemotherapy. Experimental cell research. 1998;245:350–9. [DOI] [PubMed] [Google Scholar]
  • 43.Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer cell. 2012;21:822–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. The New England journal of medicine. 2012;366:2455–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nature medicine. 2016;22:851–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Stromnes IM, Schmitt TM, Hulbert A, Brockenbrough JS, Nguyen H, Cuevas C, et al. T Cells Engineered against a Native Antigen Can Surmount Immunologic and Physical Barriers to Treat Pancreatic Ductal Adenocarcinoma. Cancer cell. 2015;28:638–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hartmann N, Giese NA, Giese T, Poschke I, Offringa R, Werner J, et al. Prevailing role of contact guidance in intrastromal T-cell trapping in human pancreatic cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014;20:3422–33. [DOI] [PubMed] [Google Scholar]
  • 48.Whatcott CJ, Diep CH, Jiang P, Watanabe A, LoBello J, Sima C, et al. Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015;21:3561–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Catenacci DV, Junttila MR, Karrison T, Bahary N, Horiba MN, Nattam SR, et al. Randomized Phase Ib/II Study of Gemcitabine Plus Placebo or Vismodegib, a Hedgehog Pathway Inhibitor, in Patients With Metastatic Pancreatic Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33:4284–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Jesus-Acosta AD, O’Dwyer PJ, Ramanathan RK, Hoff DDV, Maitra A, Rasheed Z, et al. A phase II study of vismodegib, a hedgehog (Hh) pathway inhibitor, combined with gemcitabine and nab-paclitaxel (nab-P) in patients (pts) with untreated metastatic pancreatic ductal adenocarcinoma (PDA). Journal of Clinical Oncology. 2014;32:257–.24297952 [Google Scholar]
  • 51.Gore J, Kork M. Pancreatic Cancer Stroma: Friend or Foe? Cancer cell. 2014:711–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lee JJ, Perera RM, Wang H, Wu DC, Liu XS, Han S, et al. Stromal response to Hedgehog signaling restrains pancreatic cancer progression. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E3091–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine Versus Nab-Paclitaxel/Gemcitabine in Patients With Untreated, Metastatic Pancreatic Ductal Adenocarcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2018;36:359–66. [DOI] [PubMed] [Google Scholar]
  • 54.Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer research. 1999;59:5002–11. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES